Skip to main content

Precision Medicine Based on Next-Generation Sequencing and Master Controllers

  • Reference work entry
  • First Online:
Pancreatic Cancer

Abstract

Despite decades of research and efforts at improving survival, pancreatic ductal adenocarcinoma (PDA) has become the third leading cause of cancer-related deaths in the United States. In fact, by 2020, it is projected to become the second leading cause of cancer-related deaths in the United States. Personalized, or precision, medicine has resulted in improving patient outcomes in other tumor systems. However, for pancreatic cancer patients, there are a limited number of evidence-based targeted therapeutic options that are currently available. Significant advances in DNA sequencing technology have resulted in the identification of a number of genetic mutations and the delineation of core signaling pathways important in PDA. This has subsequently resulted in an advanced understanding of the genetic drivers of the progression of this disease. Facile sequencing technology has moved the field closer to a personalized approach to treating pancreatic cancer. Improvements to the personalized therapy approach will likely result from several factors including the delivery of tumor sequencing results in a clinically relevant timeframe, the development of better targeted drugs, and perhaps a molecular-targeted approach to aspects of PDA biology beyond mutations in the deoxyribonucleic acid (DNA). These advances will allow clinicians to enroll patients in appropriate-matched clinical trials in a timely manner. In this chapter, the opportunities and limitations of a targeted, personalized approach to treating PDA will be discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 849.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 1,099.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.

    Article  PubMed  Google Scholar 

  2. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.

    Article  CAS  PubMed  Google Scholar 

  3. Winter JM, Cameron JL, Campbell KA, Arnold MA, Chang DC, Coleman J, et al. 1423 pancreaticoduodenectomies for pancreatic cancer: a single-institution experience. J Gastrointest Surg. 2006;10(9):1199–210. discussion 210-1

    Article  PubMed  Google Scholar 

  4. Von Hoff DD, Goldstein D, Renschler MF. Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N Engl J Med. 2014;370(5):479–80.

    Google Scholar 

  5. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.

    Article  CAS  PubMed  Google Scholar 

  6. Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA. The pancreas cancer microenvironment. Clin Cancer Res. 2012;18(16):4266–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chu GC, Kimmelman AC, Hezel AF, DePinho RA. Stromal biology of pancreatic cancer. J Cell Biochem. 2007;101(4):887–907.

    Article  CAS  PubMed  Google Scholar 

  8. Blanco F, Jimbo M, Wulfkuhle J, Gallagher I, Deng J, Enyenihi L, et al. The mRNA-binding protein HuR promotes hypoxia-induced chemoresistance through posttranscriptional regulation of the proto-oncogene PIM1 in pancreatic cancer cells. Oncogene. 2016;35(19):2529–41.

    Article  CAS  PubMed  Google Scholar 

  9. Prokesch RW, Schima W, Chow LC, Jeffrey RB. Multidetector CT of pancreatic adenocarcinoma: diagnostic advances and therapeutic relevance. Eur Radiol. 2003;13(9):2147–54.

    Article  PubMed  Google Scholar 

  10. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321(5897):1801–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518(7540):495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dreyer SB, Chang DK, Bailey P, Biankin AV. Pancreatic cancer genomes: implications for clinical management and therapeutic development. Clin Cancer Res. 2017;23(7):1638–46.

    Article  PubMed  Google Scholar 

  13. Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature. 2009;458(7239):719–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008;26(10):1135–45.

    Article  CAS  PubMed  Google Scholar 

  15. Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6(8):2969–72.

    CAS  PubMed  Google Scholar 

  16. Belkadi A, Bolze A, Itan Y, Cobat A, Vincent QB, Antipenko A, et al. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. Proc Natl Acad Sci. 2015;112(17):5473–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Pang AW, Macdonald JR, Yuen RK, Hayes VM, Scherer SW. Performance of high-throughput sequencing for the discovery of genetic variation across the complete size spectrum. G3 (Bethesda). 2014;4(1):63–5.

    Article  CAS  Google Scholar 

  18. Todd R, Kuo MWLWP. Gene expression profiling using laser capture microdissection. Expert Rev Mol Diagn. 2002;2(5):497–507.

    Article  CAS  PubMed  Google Scholar 

  19. Bailey P, Chang DK, Nones K, Johns AL, Patch A-M, Gingras M-C, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016. https://doi.org/10.1038/nature16965.

  20. Biankin AV, Waddell N, Kassahn KS, Gingras M-C, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491(7424):399–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17(4):500–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Campbell PJ, Yachida S, Mudie LJ, Stephens PJ, Pleasance ED, Stebbings LA, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467(7319):1109–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang L, Tsutsumi S, Kawaguchi T, Nagasaki K, Tatsuno K, Yamamoto S, et al. Whole-exome sequencing of human pancreatic cancers and characterization of genomic instability caused by MLH1 haploinsufficiency and complete deficiency. Genome Res. 2012;22(2):208–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jiao Y, Yonescu R, Offerhaus GJA, Klimstra DS, Maitra A, Eshleman JR, et al. Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. J Pathol. 2014;232(4):428–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin W-C, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744

    Google Scholar 

  27. Waddell N, Pajic M, Patch A-M, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518(7540):495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dal Molin M, Zhang M, De Wilde RF, Ottenhof NA, Rezaee N, Wolfgang CL, et al. Very long-term survival following resection for pancreatic cancer is not explained by commonly mutated genes: results of whole-exome sequencing analysis. Clin Cancer Res. 2015;21(8):1944–50.

    Article  CAS  PubMed  Google Scholar 

  29. Roberts NJ, Norris AL, Petersen GM, Bondy ML, Brand R, Gallinger S, et al. Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer. Cancer Discov. 2016;6(2):166–75.

    Article  CAS  PubMed  Google Scholar 

  30. Murphy SJ, Hart SN, Halling GC, Johnson SH, Smadbeck JB, Drucker T, et al. Integrated genomic analysis of pancreatic ductal adenocarcinomas reveals genomic rearrangement events as significant drivers of disease. Cancer Res. 2016;76(3):749–61.

    Article  CAS  PubMed  Google Scholar 

  31. Makohon-Moore AP, Zhang M, Reiter JG, Bozic I, Allen B, Kundu D, et al. Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer. Nat Genet. 2017. https://doi.org/10.1038/ng.3764.

  32. Humphris JL, Patch AM, Nones K, Bailey PJ, Johns AL, McKay S, et al. Hypermutation in pancreatic cancer. Gastroenterology. 2017;152(1):68–74.e2.

    Article  CAS  PubMed  Google Scholar 

  33. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65–71.

    Article  CAS  PubMed  Google Scholar 

  34. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW. Cancer genome landscapes. Science. 2013;339(6127):1546–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Makohon-Moore A, Iacobuzio-Donahue CA. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat Rev Cancer. 2016. https://doi.org/10.1038/nrc.2016.66.

  36. Chantrill LA, Nagrial AM, Watson C, Johns AL, Martyn-Smith M, Simpson S, et al. Precision medicine for advanced pancreas cancer: the individualized molecular pancreatic cancer therapy (IMPaCT) trial. Clin Cancer Res. 2015;21(9):2029–37.

    Article  CAS  PubMed  Google Scholar 

  37. Pishvaian MJ, Brody JR, Matrisian L, Hendifar AE, Engebretson A, Hoos WA, et al. Multi-Omic profiling (MoP) for patients (pts) with pancreatic cancer (PDA): initial results of the Know Your Tumor (KYT) initiative. Proc Am Soc Clin Oncol. 2016. https://doi.org/10.1200/jco.2016.34.4_suppl.282.

  38. Engebretson A, Brody JR, Rahib L, Matrisian L, Hendifar AE, Hoos WA, et al. The Know Your Tumor (KYT) initiative: a national program of multi-omic molecular profiling (MoP) for patients (pts) with pancreatic cancer (PDA). Proc Am Soc Clin Oncol. 2016. https://doi.org/10.1200/jco.2016.34.4_suppl.279.

  39. Mullard A. NCI-MATCH trial pushes cancer umbrella trial paradigm. Nat Rev Drug Discov. 2015;14(8):513–5.

    Article  CAS  PubMed  Google Scholar 

  40. Do K, O’Sullivan Coyne G, Chen AP. An overview of the NCI precision medicine trials – NCI MATCH and MPACT. Chin Clin Oncol. 2015;4(3):31.

    PubMed  Google Scholar 

  41. Berry DA. The brave new world of clinical cancer research: adaptive biomarker-driven trials integrating clinical practice with clinical research. Mol Oncol. 2015;9(5):951–9.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344(14):1031–7.

    Article  CAS  PubMed  Google Scholar 

  43. Maitra A, Adsay NV, Argani P, Iacobuzio-Donahue C, De Marzo A, Cameron JL, et al. Multicomponent analysis of the pancreatic adenocarcinoma progression model using a pancreatic intraepithelial neoplasia tissue microarray. Mod Pathol. 2003;16(9):902–12.

    Article  PubMed  Google Scholar 

  44. Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol. 2008;3:157–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011;11(11):761–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. McCormick F. KRAS as a Therapeutic Target. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21(8):1797–801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4(6):437–50.

    Article  CAS  PubMed  Google Scholar 

  48. Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11(3):291–302.

    Article  CAS  PubMed  Google Scholar 

  49. Seidler B, Schmidt A, Mayr U, Nakhai H, Schmid RM, Schneider G, et al. A Cre-loxP-based mouse model for conditional somatic gene expression and knockdown in vivo by using avian retroviral vectors. Proc Natl Acad Sci. 2008;105(29):10137–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Morris JP, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nat Rev Cancer. 2010;10(10):683–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ. Drugging the undruggable RAS: mission possible? Nat Rev Drug Discov. 2014;13(11):828–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Thompson H. US National Cancer Institute’s new Ras project targets an old foe. Nature medicine. 2013;19(8):949–50.

    Article  CAS  PubMed  Google Scholar 

  53. Van Cutsem E, Van De Velde H, Karasek P, Oettle H, Vervenne W, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol 2004;22(8):1430–1438.

    Google Scholar 

  54. Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, et al. Small molecule inhibition of the KRAS-PDE [delta] interaction impairs oncogenic KRAS signalling. Nature. 2013;497(7451):638.

    Article  CAS  PubMed  Google Scholar 

  55. Khvalevsky EZ, Gabai R, Rachmut IH, Horwitz E, Brunschwig Z, Orbach A, et al. Mutant KRAS is a druggable target for pancreatic cancer. Proc Natl Acad Sci. 2013;110(51):20723–8.

    Article  CAS  Google Scholar 

  56. Pecot CV, Wu SY, Bellister S, Filant J, Rupaimoole R, Hisamatsu T, et al. Therapeutic silencing of KRAS using systemically delivered siRNAs. Mol Cancer Ther. 2014;13(12):2876–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Golan T, Khvalevsky EZ, Hubert A, Gabai RM, Hen N, Segal A, et al. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget. 2015;6(27):24560.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Pratilas CA, Hanrahan AJ, Halilovic E, Persaud Y, Soh J, Chitale D, et al. Genetic predictors of MEK dependence in non–small cell lung cancer. Cancer Res. 2008;68(22):9375–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bodoky G, Timcheva C, Spigel DR, La Stella PJ, Ciuleanu TE, Pover G, et al. A phase II open-label randomized study to assess the efficacy and safety of selumetinib (AZD6244 [ARRY-142886]) versus capecitabine in patients with advanced or metastatic pancreatic cancer who have failed first-line gemcitabine therapy. Investig New Drugs. 2012;30(3):1216–23.

    Article  CAS  Google Scholar 

  60. Rinehart J, Adjei AA, LoRusso PM, Waterhouse D, Hecht JR, Natale RB, et al. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22(22):4456–62.

    Article  CAS  PubMed  Google Scholar 

  61. Infante JR, Somer BG, Park JO, Li C-P, Scheulen ME, Kasubhai SM, et al. A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur J Cancer. 2014;50(12):2072–81.

    Article  CAS  PubMed  Google Scholar 

  62. Witkiewicz AK, Borja NA, Franco J, Brody JR, Yeo CJ, Mansour J, et al. Selective impact of CDK4/6 suppression on patient-derived models of pancreatic cancer. Oncotarget. 2015;6(18):15788–801.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Franco J, Witkiewicz AK, Knudsen ES. CDK4/6 inhibitors have potent activity in combination with pathway selective therapeutic agents in models of pancreatic cancer. Oncotarget. 2014;5(15):6512–25.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Wee S, Jagani Z, Xiang KX, Loo A, Dorsch M, Yao Y-M, et al. PI3K pathway activation mediates resistance to MEK inhibitors in KRAS mutant cancers. Cancer Res. 2009;69(10):4286–93.

    Article  CAS  PubMed  Google Scholar 

  65. Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, et al. A central role for RAF→ MEK→ ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2012;2(8):685–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Alagesan B, Contino G, Guimaraes AR, Corcoran RB, Deshpande V, Wojtkiewicz GR, et al. Combined MEK and PI3K inhibition in a mouse model of pancreatic cancer. Clin Cancer Res. 2015;21(2):396–404.

    Article  CAS  PubMed  Google Scholar 

  67. Chung V, McDonough S, Philip PA, Cardin D, Wang-Gillam A, Hui L, et al. Effect of Selumetinib and MK-2206 vs Oxaliplatin and fluorouracil in patients with metastatic pancreatic cancer after prior therapy: SWOG S1115 study randomized clinical trial. JAMA Oncol. 2017;3(4):516–22.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Diep CH, Munoz RM, Choudhary A, Von Hoff DD, Han H. Synergistic effect between erlotinib and MEK inhibitors in KRAS wild-type human pancreatic cancer cells. Clin Cancer Res. 2011;17(9):2744–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kulke MH, Blaszkowsky LS, Ryan DP, Clark JW, Meyerhardt JA, Zhu AX, et al. Capecitabine plus erlotinib in gemcitabine-refractory advanced pancreatic cancer. J Clin Oncol. 2007;25(30):4787–92.

    Article  CAS  PubMed  Google Scholar 

  70. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6.

    Article  CAS  PubMed  Google Scholar 

  71. Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev Mol Cell Biol. 2001;2(10):731–7.

    Article  CAS  PubMed  Google Scholar 

  72. Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 2015;14(2):130–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Witkiewicz AK, Knudsen KE, Dicker AP, Knudsen ES. The meaning of p16ink4a expression in tumors: functional significance, clinical associations and future developments. Cell Cycle. 2011;10(15):2497–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Caldas C, Hahn SA, Da Costa LT, Redston MS, Schutte M, Seymour AB, et al. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet. 1994;8(1):27–32.

    Article  CAS  PubMed  Google Scholar 

  75. Fukushima N, Sato N, Ueki T, Rosty C, Walter KM, Wilentz RE, et al. Aberrant methylation of preproenkephalin and p16 genes in pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Am J Pathol. 2002;160(5):1573–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57(15):3126–30.

    CAS  PubMed  Google Scholar 

  77. Cicenas J, Valius M. The CDK inhibitors in cancer research and therapy. J Cancer Res Clin Oncol. 2011;137(10):1409.

    Article  CAS  PubMed  Google Scholar 

  78. Hamilton E, Infante JR. Targeting CDK4/6 in patients with cancer. Cancer Treat Rev. 2016;45:129–38.

    Article  CAS  PubMed  Google Scholar 

  79. Liu F, Korc M. Cdk4/6 inhibition induces epithelial–mesenchymal transition and enhances invasiveness in pancreatic cancer cells. Mol Cancer Ther. 2012;11(10):2138–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Heilmann AM, Perera RM, Ecker V, Nicolay BN, Bardeesy N, Benes CH, et al. CDK4/6 and IGF1 receptor inhibitors synergize to suppress the growth of p16INK4A-deficient pancreatic cancers. Cancer Res. 2014;74(14):3947–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Franco J, Witkiewicz AK, Knudsen ES. CDK4/6 inhibitors have potent activity in combination with pathway selective therapeutic agents in models of pancreatic cancer. Oncotarget. 2014;5(15):6512–25.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hruban RH, Iacobuzio-Donahue C, Wilentz RE, Goggins M, Kern SE. Molecular pathology of pancreatic cancer. Cancer J. 2000;7(4):251–8.

    Google Scholar 

  83. Khoo KH, Verma CS, Lane DP. Drugging the p53 pathway: understanding the route to clinical efficacy. Nat Rev Drug Discov. 2014;13(3):217–36.

    Article  CAS  PubMed  Google Scholar 

  84. Song G-Y, Gibson G, Haq W, Huang EC, Srivasta T, Hollstein M, et al. An MVA vaccine overcomes tolerance to human p53 in mice and humans. Cancer Immunol Immunother. 2007;56(8):1193–205.

    Article  CAS  PubMed  Google Scholar 

  85. Lehmann S, Bykov VJ, Ali D, Andrén O, Cherif H, Tidefelt U, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol. 2012;30(29):3633–9.

    Article  CAS  PubMed  Google Scholar 

  86. Izetti P, Hautefeuille A, Abujamra AL, de Farias CB, Giacomazzi J, Alemar B, et al. PRIMA-1, a mutant p53 reactivator, induces apoptosis and enhances chemotherapeutic cytotoxicity in pancreatic cancer cell lines. Investig New Drugs. 2014;32(5):783–94.

    Article  CAS  Google Scholar 

  87. Duffy MJ, Synnott NC, McGowan PM, Crown J, O’Connor D, Gallagher WM. p53 as a target for the treatment of cancer. Cancer Treat Rev. 2014;40(10):1153–60.

    Article  CAS  PubMed  Google Scholar 

  88. Yu X, Narayanan S, Vazquez A, Carpizo DR. Small molecule compounds targeting the p53 pathway: are we finally making progress? Apoptosis. 2014;19(7):1055–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Hahn SA, Greenhalf B, Ellis I, Sina-Frey M, Rieder H, Korte B, et al. BRCA2 germline mutations in familial pancreatic carcinoma. J Natl Cancer Inst. 2003;95(3):214–21.

    Article  CAS  PubMed  Google Scholar 

  90. van der Heijden MS, Yeo CJ, Hruban RH, Kern SE. Fanconi anemia gene mutations in young-onset pancreatic cancer. Cancer Res. 2003;63(10):2585–8.

    PubMed  Google Scholar 

  91. Van der Heijden MS, Brody JR, Gallmeier E, Cunningham SC, Dezentje DA, Shen D, et al. Functional defects in the fanconi anemia pathway in pancreatic cancer cells. Am J Pathol. 2004;165(2):651–7.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Gallmeier E, Calhoun ES, Rago C, Brody JR, Cunningham SC, Hucl T, et al. Targeted disruption of FANCC and FANCG in human cancer provides a preclinical model for specific therapeutic options. Gastroenterology. 2006;130(7):2145–54.

    Article  CAS  PubMed  Google Scholar 

  93. Murphy SJ, Hart SN, Lima JF, Kipp BR, Klebig M, Winters JL, et al. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology. 2013;145(5):1098–109. e1.

    Article  CAS  PubMed  Google Scholar 

  94. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med. 2012;366(15):1382–92.

    Article  CAS  PubMed  Google Scholar 

  95. Golan T, Kanji Z, Epelbaum R, Devaud N, Dagan E, Holter S, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer. 2014;111(6):1132–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ashworth A. A synthetic lethal therapeutic approach: poly (ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol. 2008;26(22):3785–90.

    Article  CAS  PubMed  Google Scholar 

  97. Benafif S, Hall M. An update on PARP inhibitors for the treatment of cancer. Onco Targets Ther. 2015;8:519.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Kaelin WG. The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer. 2005;5(9):689–98.

    Article  CAS  PubMed  Google Scholar 

  99. Villarroel MC, Rajeshkumar N, Garrido-Laguna I, De Jesus-Acosta A, Jones S, Maitra A, et al. Personalizing cancer treatment in the age of global genomic analyses: PALB2 gene mutations and the response to DNA damaging agents in pancreatic cancer. Mol Cancer Ther. 2011;10(1):3–8.

    Article  CAS  PubMed  Google Scholar 

  100. Pishvaian MJ, Wang H, Zhuang T, He AR, Hwang JJ, Hankin A, et al. A phase I/II study of ABT-888 in combination with 5-fluorouracil (5-FU) and oxaliplatin (Ox) in patients with metastatic pancreatic cancer (MPC). Proc Am Soc Clin Oncol. 2013.

    Google Scholar 

  101. Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmana J, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol Off J Am Soc Clin Oncol. 2015;33(3):244–50.

    Article  CAS  Google Scholar 

  102. Weber AM, Ryan AJ. ATM and ATR as therapeutic targets in cancer. Pharmacol Ther. 2015;149:124–38.

    Article  CAS  PubMed  Google Scholar 

  103. Karnitz LM, Zou L. Molecular pathways: targeting ATR in cancer therapy. Clin Cancer Res. 2015;21(21):4780–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Prevo R, Fokas E, Reaper PM, Charlton PA, Pollard JR, McKenna WG, et al. The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther. 2012;13(11):1072–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bang Y-J, Im S-A, Lee K-W, Cho JY, Song E-K, Lee KH, et al. Randomized, double-blind phase II trial with prospective classification by ATM protein level to evaluate the efficacy and tolerability of olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer. J Clin Oncol. 2015;33(33):3858–65.

    Article  CAS  PubMed  Google Scholar 

  106. Kadoch C, Hargreaves DC, Hodges C, Elias L, Ho L, Ranish J, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45(6):592–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Wilson BG, Roberts CW. SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 2011;11(7):481–92.

    Article  CAS  PubMed  Google Scholar 

  108. Wu C. Chromatin remodeling and the control of gene expression. J Biol Chem. 1997;272(45):28171–4.

    Article  CAS  PubMed  Google Scholar 

  109. Narlikar GJ, Fan H-Y, Kingston RE. Cooperation between complexes that regulate chromatin structure and transcription. Cell. 2002;108(4):475–87.

    Article  CAS  PubMed  Google Scholar 

  110. Khursheed M, Kolla J, Kotapalli V, Gupta N, Gowrishankar S, Uppin S, et al. ARID1B, a member of the human SWI/SNF chromatin remodeling complex, exhibits tumour-suppressor activities in pancreatic cancer cell lines. Br J Cancer. 2013;108(10):2056–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Numata M, Morinaga S, Watanabe T, Tamagawa H, Yamamoto N, Shiozawa M, et al. The clinical significance of SWI/SNF complex in pancreatic cancer. Int J Oncol. 2013;42(2):403–10.

    Article  PubMed  Google Scholar 

  112. Knudsen ES, O’Reilly EM, Brody JR, Witkiewicz AK. Genetic diversity of pancreatic ductal adenocarcinoma and opportunities for precision medicine. Gastroenterology. 2016;150(1):48–63.

    Article  PubMed  Google Scholar 

  113. Shen J, Peng Y, Wei L, Zhang W, Yang L, Lan L, et al. ARID1A deficiency impairs the DNA damage checkpoint and sensitizes cells to PARP inhibitors. Cancer Discov. 2015;5(7):752–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Williamson CT, Miller R, Pemberton HN, Jones SE, Campbell J, Konde A, et al. ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A. Nat Commun. 2016;7:13837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Truty MJ, Urrutia R. Basics of TGF-ß and pancreatic cancer. Pancreatology. 2007;7(5–6):423–35.

    Article  CAS  PubMed  Google Scholar 

  116. Ijichi H, Ikenoue T, Kato N, Mitsuno Y, Togo G, Kato J, et al. Systematic analysis of the TGF-β-Smad signaling pathway in gastrointestinal cancer cells. Biochem Biophys Res Commun. 2001;289(2):350–7.

    Article  CAS  PubMed  Google Scholar 

  117. Hahn SA, Schutte M, Hoque A. Moskaluk CA. DCP4, a candidate tumor supressor gene at human chromosome 18q21. 1. Science. 1996;271(5247):350.

    Article  CAS  PubMed  Google Scholar 

  118. Yoo J, Ghiassi M, Jirmanova L, Balliet AG, Hoffman B, Fornace AJ, et al. Transforming growth factor-β-induced apoptosis is mediated by Smad-dependent expression of GADD45b through p38 activation. J Biol Chem. 2003;278(44):43001–7.

    Article  CAS  PubMed  Google Scholar 

  119. Embuscado EE, Laheru D, Ricci F, Yun KJ, de Boom Witzel S, Seigel A, et al. Immortalizing the complexity of cancer metastasis: genetic features of lethal metastatic pancreatic cancer obtained from rapid autopsy. Cancer Biol Ther. 2005;4(5):548–54.

    Article  CAS  PubMed  Google Scholar 

  120. Iacobuzio-Donahue CA, Fu B, Yachida S, Luo M, Abe H, Henderson CM, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol. 2009;27(11):1806–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Winter JM, Tang LH, Klimstra DS, Liu W, Linkov I, Brennan MF, et al. Failure patterns in resected pancreas adenocarcinoma: lack of predicted benefit to SMAD4 expression. Ann Surg. 2013;258(2):331.

    Article  PubMed  Google Scholar 

  122. Colak S, ten Dijke P. Targeting TGF-β Signaling in cancer. Trends Cancer. 2017;3:56–71.

    Article  PubMed  Google Scholar 

  123. Neuzillet C, de Gramont A, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, et al. Perspectives of TGF-beta inhibition in pancreatic and hepatocellular carcinomas. Oncotarget. 2014;5(1):78–94.

    Article  PubMed  Google Scholar 

  124. Melisi D, Garcia-Carbonero R, Macarulla T, Pezet D, Deplanque G, Fuchs M, et al. A phase II, double-blind study of galunisertib+ gemcitabine (GG) vs gemcitabine+ placebo (GP) in patients (pts) with unresectable pancreatic cancer (PC). Proc Am Soc Clin Oncol. 2016.

    Google Scholar 

  125. Blackford A, Serrano OK, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15(14):4674–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Tatarian T, Winter JM. Genetics of pancreatic cancer and its implications on therapy. Surg Clin N Am. 2016;96(6):1207–21.

    Article  PubMed  Google Scholar 

  127. Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the US intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18(5):1319–26.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Network CGA. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487(7407):330–7.

    Article  CAS  Google Scholar 

  129. White BD, Chien AJ, Dawson DW. Dysregulation of Wnt/β-catenin signaling in gastrointestinal cancers. Gastroenterology. 2012;142(2):219–32.

    Article  CAS  PubMed  Google Scholar 

  130. Jiang X, Hao H-X, Growney JD, Woolfenden S, Bottiglio C, Ng N, et al. Inactivating mutations of RNF43 confer Wnt dependency in pancreatic ductal adenocarcinoma. Proc Natl Acad Sci. 2013;110(31):12649–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Zhang Y, Morris JP, Yan W, Schofield HK, Gurney A, Simeone DM, et al. Canonical wnt signaling is required for pancreatic carcinogenesis. Cancer Res. 2013;73(15):4909–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Jiang X, Charlat O, Zamponi R, Yang Y, Cong F. Dishevelled promotes Wnt receptor degradation through recruitment of ZNRF3/RNF43 E3 ubiquitin ligases. Mol Cell. 2015;58(3):522–33.

    Article  CAS  PubMed  Google Scholar 

  133. Liu J, Pan S, Hsieh MH, Ng N, Sun F, Wang T, et al. Targeting Wnt-driven cancer through the inhibition of porcupine by LGK974. Proc Natl Acad Sci. 2013;110(50):20224–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Ranganathan P, Weaver KL, Capobianco AJ. Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer. 2011;11(5):338–51.

    Article  CAS  PubMed  Google Scholar 

  135. Gao J, Long B, Wang Z. Role of notch signaling pathway in pancreatic cancer. Am J Cancer Res. 2017;7(2):173–86.

    PubMed  PubMed Central  Google Scholar 

  136. De La OJ, Murtaugh LC. Notch and Kras in pancreatic cancer: at the crossroads of mutation, differentiation and signaling. Cell Cycle. 2009;8(12):1860–4.

    Article  Google Scholar 

  137. Thomas MM, Zhang Y, Mathew E, Kane KT, Maillard I, Pasca di Magliano M. Epithelial notch signaling is a limiting step for pancreatic carcinogenesis. BMC Cancer. 2014;14:862.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, et al. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69(6):2400–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Kabashima-Niibe A, Higuchi H, Takaishi H, Masugi Y, Matsuzaki Y, Mabuchi Y, et al. Mesenchymal stem cells regulate epithelial-mesenchymal transition and tumor progression of pancreatic cancer cells. Cancer Sci. 2013;104(2):157–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Du X, Zhao YP, Zhang TP, Zhou L, Chen G, Wang TX, et al. Alteration of the intrinsic apoptosis pathway is involved in notch-induced chemoresistance to gemcitabine in pancreatic cancer. Arch Med Res. 2014;45(1):15–20.

    Article  CAS  PubMed  Google Scholar 

  141. Doucas H, Mann CD, Sutton CD, Garcea G, Neal CP, Berry DP, et al. Expression of nuclear Notch3 in pancreatic adenocarcinomas is associated with adverse clinical features, and correlates with the expression of STAT3 and phosphorylated Akt. J Surg Oncol. 2008;97(1):63–8.

    Article  PubMed  Google Scholar 

  142. Mann CD, Bastianpillai C, Neal CP, Masood MM, Jones DJ, Teichert F, et al. Notch3 and HEY-1 as prognostic biomarkers in pancreatic adenocarcinoma. PLoS One. 2012;7(12):e51119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Mizuma M, Rasheed ZA, Yabuuchi S, Omura N, Campbell NR, de Wilde RF, et al. The gamma secretase inhibitor MRK-003 attenuates pancreatic cancer growth in preclinical models. Mol Cancer Ther. 2012;11(9):1999–2009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Palagani V, Bozko P, El Khatib M, Belahmer H, Giese N, Sipos B, et al. Combined inhibition of notch and JAK/STAT is superior to monotherapies and impairs pancreatic cancer progression. Carcinogenesis. 2014;35(4):859–66.

    Article  CAS  PubMed  Google Scholar 

  145. Yabuuchi S, Pai SG, Campbell NR, de Wilde RF, De Oliveira E, Korangath P, et al. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013;335(1):41–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Cook N, Frese KK, Bapiro TE, Jacobetz MA, Gopinathan A, Miller JL, et al. Gamma secretase inhibition promotes hypoxic necrosis in mouse pancreatic ductal adenocarcinoma. J Exp Med. 2012;209(3):437–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shih Ie M, Wang TL. Notch signaling, gamma-secretase inhibitors, and cancer therapy. Cancer Res. 2007;67(5):1879–82.

    Article  PubMed  CAS  Google Scholar 

  148. De Jesus-Acosta A, Laheru D, Maitra A, Arcaroli J, Rudek MA, Dasari A, et al. A phase II study of the gamma secretase inhibitor RO4929097 in patients with previously treated metastatic pancreatic adenocarcinoma. Investig New Drugs. 2014;32(4):739–45.

    Article  CAS  Google Scholar 

  149. Yen W-C, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, et al. Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21(9):2084–95.

    Article  CAS  PubMed  Google Scholar 

  150. O’Reilly EM, Smith LS, Bendell JC, Strickler JH, Zalupski M, Gluck W, et al. Final results of phase Ib of anticancer stem cell antibody tarextumab (OMP-59R5, TRXT, anti-Notch 2/3) in combination with nab-paclitaxel and gemcitabine (Nab-P+ Gem) in patients (pts) with untreated metastatic pancreatic cancer (mPC). Proc Am Soc Clin Oncol. 2015. https://doi.org/10.1200/jco.2015.33.3_suppl.278.

  151. O’REILLY EM, Sahai V, Bendell JC, BULLOCK A, LOCONTE N, Hatoum H, et al. Results of a randomized phase 2 trial of an anti-notch 2/3, Tarextumab (OMP-59R5, TRXT, anti-notch 2/3), in combination with Nab-paclitaxel and Gemcitabine (Nab-P+ Gem) in patients (pts) with untreated metastatic pancreatic cancer (mPC). Proc Am Soc Clin Oncol. 2017. https://doi.org/10.1200/JCO.2017.35.4_suppl.279.

  152. Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008;22(18):2454–72.

    Article  CAS  PubMed  Google Scholar 

  153. Jiang J, Hui CC. Hedgehog signaling in development and cancer. Dev Cell. 2008;15(6):801–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Yang L, Xie G, Fan Q, Xie J. Activation of the hedgehog-signaling pathway in human cancer and the clinical implications. Oncogene. 2010;29(4):469–81.

    Article  PubMed  CAS  Google Scholar 

  155. Nakashima H, Nakamura M, Yamaguchi H, Yamanaka N, Akiyoshi T, Koga K, et al. Nuclear factor-kappaB contributes to hedgehog signaling pathway activation through sonic hedgehog induction in pancreatic cancer. Cancer Res. 2006;66(14):7041–9.

    Article  CAS  PubMed  Google Scholar 

  156. Kasperczyk H, Baumann B, Debatin KM, Fulda S. Characterization of sonic hedgehog as a novel NF-kappaB target gene that promotes NF-kappaB-mediated apoptosis resistance and tumor growth in vivo. FASEB J. 2009;23(1):21–33.

    Article  CAS  PubMed  Google Scholar 

  157. Ling J, Kang Y, Zhao R, Xia Q, Lee DF, Chang Z, et al. KrasG12D-induced IKK2/beta/NF-kappaB activation by IL-1alpha and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(1):105–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature. 2003;425(6960):851–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Apelqvist Å, Ahlgren U, Edlund H. Sonic hedgehog directs specialised mesoderm differentiation in the intestine and pancreas. Curr Biol. 1997;7(10):801–4.

    Article  CAS  PubMed  Google Scholar 

  160. di Magliano MP, Sekine S, Ermilov A, Ferris J, Dlugosz AA, Hebrok M. Hedgehog/Ras interactions regulate early stages of pancreatic cancer. Genes Dev. 2006;20(22):3161–73.

    Article  CAS  Google Scholar 

  161. Nolan-Stevaux O, Lau J, Truitt ML, Chu GC, Hebrok M, Fernández-Zapico ME, et al. GLI1 is regulated through smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev. 2009;23(1):24–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene. 2009;28(40):3513–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Bailey JM, Swanson BJ, Hamada T, Eggers JP, Singh PK, Caffery T, et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res. 2008;14(19):5995–6004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, Mullendore M, et al. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67(5):2187–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Macarulla T, Tabernero J, Palmer DH, Sharma S, KH Y, Sellami DB, et al. A phase Ib dose escalation, safety, and tolerability study of sonidegib in combination with gemcitabine in patients with locally advanced or metastatic pancreatic adenocarcinoma. Proc Am Soc Clin Oncol. 2016. https://doi.org/10.1200/jco.2016.34.4_suppl.371.

  167. Singh BN, Fu J, Srivastava RK, Shankar S. Hedgehog signaling antagonist GDC-0449 (Vismodegib) inhibits pancreatic cancer stem cell characteristics: molecular mechanisms. PLoS One. 2011;6(11):e27306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Kim EJ, Sahai V, Abel EV, Griffith KA, Greenson JK, Takebe N, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin Cancer Res. 2014. https://doi.org/10.1158/1078-0432.CCR-14-1269.

  169. Hidalgo M, Cooray P, Jameson MB, Carrato A, Parnis F, Jeffery M, et al. A phase Ib study of the anti-cancer stem cell agent demcizumab (DEM) & gemcitabine (GEM)+/−paclitaxel protein bound particles (nab-paclitaxel) in pts with pancreatic cancer. Proc Am Soc Clin Oncol. 2015.

    Google Scholar 

  170. Feldmann G, Habbe N, Dhara S, Bisht S, Alvarez H, Fendrich V, et al. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut. 2008;57(10):1420–30.

    Article  CAS  PubMed  Google Scholar 

  171. Liu X, Krawczyk E, Suprynowicz FA, Palechor-Ceron N, Yuan H, Dakic A, et al. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat Protoc. 2017;12(2):439–51.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  172. Beglyarova N, Banina E, Zhou Y, Mukhamadeeva R, Andrianov G, Bobrov E, et al. Screening of conditionally reprogrammed patient-derived carcinoma cells identifies ERCC3–MYC interactions as a target in pancreatic cancer. Clin Cancer Res. 2016;22(24):6153–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van Den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.

    Article  CAS  PubMed  Google Scholar 

  174. van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  175. Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256–62.

    Article  CAS  PubMed  Google Scholar 

  176. Huang L, Holtzinger A, Jagan I, BeGora M, Lohse I, Ngai N, et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell-and patient-derived tumor organoids. Nat Med. 2015;21(11):1364–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Boj SF, Hwang C-I, Baker LA, Chio IIC, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1):324–38.

    Article  CAS  PubMed  Google Scholar 

  178. Baker LA, Tiriac H, Clevers H, Tuveson DA. Modeling pancreatic cancer with organoids. Trends Cancer. 2016;2(4):176–90.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Drost J, Van Jaarsveld RH, Ponsioen B, Zimberlin C, Van Boxtel R, Buijs A, et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature. 2015;521(7550):43–7.

    Article  CAS  PubMed  Google Scholar 

  180. Ribas A, Kefford R, Marshall MA, Punt CJ, Haanen JB, Marmol M, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31(5):616–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti–PD-1) in melanoma. N Engl J Med. 2013;369(2):134–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Riley JL. Combination checkpoint blockade–taking melanoma immunotherapy to the next level. The New England journal of medicine. 2013;369(2):187–9.

    Article  CAS  PubMed  Google Scholar 

  183. Keene JD. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet. 2007;8(7):533–43.

    Article  CAS  PubMed  Google Scholar 

  184. Day D, Tuite MF. Post-transcriptional gene regulatory mechanisms in eukaryotes: an overview. J Endocrinol. 1998;157(3):361–71.

    Article  CAS  PubMed  Google Scholar 

  185. Glisovic T, Bachorik JL, Yong J, Dreyfuss G. RNA-binding proteins and post-transcriptional gene regulation. FEBS Lett. 2008;582(14):1977–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Audic Y, Hartley RS. Post-transcriptional regulation in cancer. Biol Cell. 2004;96(7):479–98.

    Article  CAS  PubMed  Google Scholar 

  187. Jewer M, Findlay SD, Postovit L-M. Post-transcriptional regulation in cancer progression. J Cell Comm Signal. 2012;6(4):233–48.

    Article  Google Scholar 

  188. Dreyfuss G, Kim VN, Kataoka N. Messenger-RNA-binding proteins and the messages they carry. Nat Rev Mol Cell Biol. 2002;3(3):195–205.

    Article  CAS  PubMed  Google Scholar 

  189. Kechavarzi B, Janga SC. Dissecting the expression landscape of RNA-binding proteins in human cancers. Genome Biol. 2014;15(1):R14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Srikantan S, Gorospe M. HuR function in disease. Front Biosci. 2012;17:189.

    Article  CAS  Google Scholar 

  191. Brennan CM, Gallouzi I-E, Steitz JA. Protein ligands to HuR modulate its interaction with target mRNAs in vivo. J Cell Biol. 2000;151(1):1–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Kim HH, Abdelmohsen K, Gorospe M. Regulation of HuR by DNA damage response kinases. J Nucleic Acids. 2010;2010:981487.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Jimbo M, Blanco FF, Huang Y-H, Telonis AG, Screnci BA, Cosma GL, et al. Targeting the mRNA-binding protein HuR impairs malignant characteristics of pancreatic ductal adenocarcinoma cells. Oncotarget. 2015;6(29):27312.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Lal S, Burkhart RA, Beeharry N, Bhattacharjee V, Londin ER, Cozzitorto JA, et al. HuR posttranscriptionally regulates WEE1: implications for the DNA damage response in pancreatic cancer cells. Cancer Res. 2014;74(4):1128–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Pineda DM, Rittenhouse DW, Valley CC, Cozzitorto JA, Burkhart RA, Leiby B, et al. HuR’s post-transcriptional regulation of death receptor 5 in pancreatic cancer cells. Cancer Biol Ther. 2012;13(10):946–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Burkhart RA, Pineda DM, Chand SN, Romeo C, Londin ER, Karoly ED, et al. HuR is a post-transcriptional regulator of core metabolic enzymes in pancreatic cancer. RNA Biol. 2013;10(8):1312–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Lal S, Cheung EC, Zarei M, Preet R, Chand SN, Mambelli-Lisboa NC, et al. CRISPR knockout of the HuR gene causes a xenograft lethal phenotype. Mol Cancer Res. 2017;15:696–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Blanco FF, Preet R, Aguado A, Vishwakarma V, Stevens LE, Vyas A, et al. Impact of HuR inhibition by the small molecule MS-444 on colorectal cancer cell tumorigenesis. Oncotarget. 2016;7:74043–58.

    PubMed  PubMed Central  Google Scholar 

  199. Kaur K, Wu X, Fields JK, Johnson DK, Lan L, Pratt M, et al. The fungal natural product azaphilone-9 binds to HuR and inhibits HuR-RNA interaction in vitro. PLoS One. 2017;12(4):e0175471.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Richards NG, Rittenhouse DW, Freydin B, Cozzitorto JA, Grenda D, Rui H, et al. HuR status is a powerful marker for prognosis and response to gemcitabine-based chemotherapy for resected pancreatic ductal adenocarcinoma patients. Ann Surg. 2010;252(3):499–506.

    PubMed  Google Scholar 

  201. Tatarian T, JiangW, Leiby BE, Grigoli A, Jimbo M, Dabbish N, et al. Cytoplasmic HuR Status Predicts Disease-free Survival in Resected Pancreatic Cancer: A Post-hoc Analysis From the International Phase III ESPAC-3 Clinical Trial. Ann Surg. 2017. https://doi.org/10.1097/SLA.0000000000002088

  202. Suvà ML, Riggi N, Bernstein BE. Epigenetic reprogramming in cancer. Science. 2013;339(6127):1567–70.

    Article  PubMed  CAS  Google Scholar 

  203. Deer EL, González-Hernández J, Coursen JD, Shea JE, Ngatia J, Scaife CL, et al. Phenotype and genotype of pancreatic cancer cell lines. Pancreas. 2010;39(4):425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. McDonald OG, Li X, Saunders T, Tryggvadottir R, Mentch SJ, Warmoes MO, et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nature genetics. 2017;49(3):367–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Millward M, Price T, Townsend A, Sweeney C, Spencer A, Sukumaran S, et al. Phase 1 clinical trial of the novel proteasome inhibitor marizomib with the histone deacetylase inhibitor vorinostat in patients with melanoma, pancreatic and lung cancer based on in vitro assessments of the combination. Investig New Drugs. 2012;30(6):2303–17.

    Article  CAS  Google Scholar 

  206. Kumagai T, Wakimoto N, Yin D, Gery S, Kawamata N, Takai N, et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (Vorinostat, SAHA) profoundly inhibits the growth of human pancreatic cancer cells. Int J Cancer. 2007;121(3):656–65.

    Article  CAS  PubMed  Google Scholar 

  207. Lee HS, Park SB, Kim SA, Kwon SK, Cha H, Lee DY, et al. A novel HDAC inhibitor, CG200745, inhibits pancreatic cancer cell growth and overcomes gemcitabine resistance. Sci Rep. 2017;7:41615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Kristensen VN, Lingjærde OC, Russnes HG, Vollan HKM, Frigessi A, Børresen-Dale A-L. Principles and methods of integrative genomic analyses in cancer. Nat Rev Cancer. 2014;14(5):299–313.

    Article  CAS  PubMed  Google Scholar 

  209. Pishvaian MJ, Matrisian L, Hendifar AE, Engebretson A, Rahib L, Hoos WA, et al. Preliminary observations of blood-based (BB) molecular testing in a subset of patients with pancreatic cancer (PDA) participating in the Know Your Tumor (KYT) initiative. Proc Am Soc Clin Oncol. 2016;34:268.

    Article  Google Scholar 

  210. Bender RJ, Halverson D, Mason K, Luo L, Brody JR, Rahib L, et al. Molecular biomarkers as predictors of patient survival in pancreatic adenocarcinoma (PDA): An analysis of the Know Your Tumor initiative (KYT). Journal of Clinical Oncology. 2017;35(4_suppl):278.

    Article  Google Scholar 

  211. Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135(5):1110–8.

    Article  CAS  PubMed  Google Scholar 

  212. Narayan G, Goparaju C, Arias-Pulido H, Kaufmann AM, Schneider A, Dürst M, et al. Promoter hypermethylation-mediated inactivation of multiple Slit-Robo pathway genes in cervical cancer progression. Mol Cancer. 2006;5(1):16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Xian J, Clark KJ, Fordham R, Pannell R, Rabbitts TH, Rabbitts PH. Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc Natl Acad Sci. 2001;98(26):15062–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Dumartin L, Quemener C, Laklai H, Herbert J, Bicknell R, Bousquet C, et al. Netrin-1 mediates early events in pancreatic adenocarcinoma progression, acting on tumor and endothelial cells. Gastroenterology. 2010;138(4):1595–606. e8.

    Article  CAS  PubMed  Google Scholar 

  215. Ricci F, Kern SE, Hruban RH, Iacobuzio-Donahue CA. Stromal responses to carcinomas of the pancreas: juxtatumoral gene expression conforms to the infiltrating pattern and not the biologic subtype. Cancer Biol Ther. 2005;4(3):302–7.

    Article  CAS  PubMed  Google Scholar 

  216. Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68(3):918–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Mahadevan D, Von Hoff DD. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2007;6(4):1186–97.

    Article  CAS  PubMed  Google Scholar 

  218. Kong X, Li L, Li Z, Xie K. Targeted destruction of the orchestration of the pancreatic stroma and tumor cells in pancreatic cancer cases: molecular basis for therapeutic implications. Cytokine Growth Factor Rev. 2012;23(6):343–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Sherman MH, Ruth TY, Tseng TW, Sousa CM, Liu S, Truitt ML, et al. Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc Natl Acad Sci. 2017;114:1129–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Apte M, Pirola RC, Wilson JS. Pancreatic stellate cell: physiologic role, role in fibrosis and cancer. Curr Opin Gastroenterol. 2015;31(5):416–23.

    Article  CAS  PubMed  Google Scholar 

  221. Sherman MH, Ruth TY, Engle DD, Ding N, Atkins AR, Tiriac H, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell. 2014;159(1):80–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Vogelmann R, Ruf D, Wagner M, Adler G, Menke A. Effects of fibrogenic mediators on the development of pancreatic fibrosis in a TGF-β1 transgenic mouse model. Am J Physiol –Gastrointest Liver Physiol. 2001;280(1):G164–G72.

    Article  CAS  PubMed  Google Scholar 

  223. Löhr M, Schmidt C, Ringel J, Kluth M, Müller P, Nizze H, et al. Transforming growth factor-β1 induces desmoplasia in an experimental model of human pancreatic carcinoma. Cancer Res. 2001;61(2):550–5.

    PubMed  Google Scholar 

  224. Bever KM, Sugar EA, Bigelow E, Sharma R, Laheru D, Wolfgang CL, et al. The prognostic value of stroma in pancreatic cancer in patients receiving adjuvant therapy. HPB. 2015;17(4):292–8.

    Article  PubMed  Google Scholar 

  225. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Tempero MA, Coussens LM, Fong L, Manges R, Singh P, Li Y, et al. A randomized, multicenter, double-blind, placebo-controlled study of the Bruton tyrosine kinase inhibitor, ibrutinib, versus placebo in combination with nab-paclitaxel and gemcitabine in the first-line treatment of patients with metastatic pancreatic adenocarcinoma (RESOLVE). Journal of Clinical Oncology. 2016;34(4_suppl):TPS483-TPS.

    Article  Google Scholar 

  227. Borazanci EH, Hong DS, Gutierrez M, Rasco DW, Reid TR, Veeder MH, et al. Ibrutinib + durvalumab (MEDI4736) in patients (pts) with relapsed or refractory (R/R) pancreatic adenocarcinoma (PAC): A phase Ib/II multicenter study. Journal of Clinical Oncology. 2016;34(4_suppl):TPS484-TPS.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jonathan R. Brody .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC, part of Springer Nature

About this entry

Check for updates. Verify currency and authenticity via CrossMark

Cite this entry

Dukleska, K., Yeo, C.J., Pishvaian, M.J., Brody, J.R. (2018). Precision Medicine Based on Next-Generation Sequencing and Master Controllers. In: Neoptolemos, J., Urrutia, R., Abbruzzese, J., Büchler, M. (eds) Pancreatic Cancer. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-7193-0_71

Download citation

Publish with us

Policies and ethics